1
|
Zhu X, Lu M, Li WX, Lin L, Liu Y, Zhou J, Shang J, Shi X, Lu J, Xing J, Zhang M, Zhao S, Zhao D. HuMSCs-derived exosomal YBX1 participates in oxidative damage repair in granulosa cells by stabilizing COX5B mRNA in an m5C-dependent manner. Int J Biol Macromol 2025; 310:143288. [PMID: 40253045 DOI: 10.1016/j.ijbiomac.2025.143288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 03/06/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Mitochondrial dysfunction and cell senescence are triggered by reactive oxygen species (ROS) in granulosa cells (GCs), leading to premature ovarian insufficiency (POI). Human umbilical cord mesenchymal stem cell-derived exosome (HuMSCs-Ex, H-Ex)-based treatments have been shown to alleviate ROS-induced POI, but knowledge about the underlying therapeutic mechanisms is limited. Here, we observed that the 5-methylcytosine (m5C) RNA methyltransferase tRNA aspartic acid methyltransferase 1 (TRDMT1) promoted the translation of COX subunit 5B (COX5B) in a manner dependent on its catalytic activity and downstream m5C reader Y-box binding protein 1 (YBX1), which was decreased in prematurely senescent GCs but abundant in H-Ex. Mechanistically, YBX1 released by H-Ex recognizes the TRDMT1-mediated m5C modification of COX5B and directly binds to COX5BC-153 via LYS-92, thereby reducing ROS accumulation and improving mitochondrial function in GCs under oxidative stress, providing new insights into the theoretical basis for the great clinical potential of H-Ex in the treatment of POI.
Collapse
Affiliation(s)
- Xiaolan Zhu
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China.
| | - Minjun Lu
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China; Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China
| | - Wen-Xin Li
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China; Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China
| | - Li Lin
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China; Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China
| | - Yueqin Liu
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China
| | - Jiamin Zhou
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China; Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China
| | - Junyu Shang
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China; Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China
| | - Xuyan Shi
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China; Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China
| | - Jingjing Lu
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China; Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China
| | - Jie Xing
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China; Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China
| | - Mengxue Zhang
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China; Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China
| | - Shijie Zhao
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China; Department of Central Laboratory, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China
| | - Dan Zhao
- Department of Reproductive Medical Center, Fourth Affiliated Hospital of Jiangsu University (Zhenjiang Maternity and Child Health Care Hospital), Zhenjiang, China
| |
Collapse
|
2
|
Choudhury FK, Premkumar V, Zecha J, Boyd J, Gaynor AS, Guo Z, Martin T, Cimbro R, Allman EL, Hess S. Multiomics Characterization of a Less Invasive Microfluidic-Based Cell Sorting Technique. J Proteome Res 2024; 23:3096-3107. [PMID: 38417049 PMCID: PMC11301668 DOI: 10.1021/acs.jproteome.3c00773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/27/2024] [Accepted: 02/12/2024] [Indexed: 03/01/2024]
Abstract
Fluorescence-activated cell sorting (FACS) is a specialized technique to isolate specific cell subpopulations with a high level of recovery and accuracy. However, the cell sorting procedure can impact the viability and metabolic state of cells. Here, we performed a comparative study and evaluated the impact of traditional high-pressure charged droplet-based and microfluidic chip-based sorting on the metabolic and phosphoproteomic profile of different cell types. While microfluidic chip-based sorted cells more closely resembled the unsorted control group for most cell types tested, the droplet-based sorted cells showed significant metabolic and phosphoproteomic alterations. In particular, greater changes in redox and energy status were present in cells sorted with the droplet-based cell sorter along with larger shifts in proteostasis. 13C-isotope tracing analysis on cells recovering postsorting revealed that the sorter-induced suppression of mitochondrial TCA cycle activity recovered faster in the microfluidic chip-based sorted group. Apart from this, amino acid and lipid biosynthesis pathways were suppressed in sorted cells, with minimum impact and faster recovery in the microfluidic chip-based sorted group. These results indicate microfluidic chip-based sorting has a minimum impact on metabolism and is less disruptive compared to droplet-based sorting.
Collapse
Affiliation(s)
- Feroza K. Choudhury
- Dynamic
Omics, Centre for Genomics Research (CGR), Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Viji Premkumar
- Dynamic
Omics, Centre for Genomics Research (CGR), Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Jana Zecha
- Dynamic
Omics, Centre for Genomics Research (CGR), Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Jonathan Boyd
- Dynamic
Omics, Centre for Genomics Research (CGR), Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Andrew S. Gaynor
- Oncology
Cell Therapy, ICC, Oncology R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Zengli Guo
- Biologics
Engineering, Cell Therapeutics and Viral Technologies, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Tom Martin
- Biologics
Engineering, Oncology TTD, ADC, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Raffaello Cimbro
- Dynamic
Omics, Centre for Genomics Research (CGR), Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, United Kingdom
| | - Erik L. Allman
- Dynamic
Omics, Centre for Genomics Research (CGR), Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Sonja Hess
- Dynamic
Omics, Centre for Genomics Research (CGR), Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| |
Collapse
|
3
|
Wang SY, Wang YX, Shen A, Yang XQ, Liang CC, Huang RJ, Jian R, An N, Xiao YL, Wang LS, Zhao Y, Lin C, Wang CP, Yuan ZP, Yuan SQ. Construction of a gene model related to the prognosis of patients with gastric cancer receiving immunotherapy and exploration of COX7A1 gene function. Eur J Med Res 2024; 29:180. [PMID: 38494472 PMCID: PMC11337786 DOI: 10.1186/s40001-024-01783-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 03/10/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND GC is a highly heterogeneous tumor with different responses to immunotherapy, and the positive response depends on the unique interaction between the tumor and the tumor microenvironment (TME). However, the currently available methods for prognostic prediction are not satisfactory. Therefore, this study aims to construct a novel model that integrates relevant gene sets to predict the clinical efficacy of immunotherapy and the prognosis of GC patients based on machine learning. METHODS Seven GC datasets were collected from the Gene Expression Omnibus (GEO) database, The Cancer Genome Atlas (TCGA) database and literature sources. Based on the immunotherapy cohort, we first obtained a list of immunotherapy related genes through differential expression analysis. Then, Cox regression analysis was applied to divide these genes with prognostic significancy into protective and risky types. Then, the Single Sample Gene Set Enrichment Analysis (ssGSEA) algorithm was used to score the two categories of gene sets separately, and the scores differences between the two gene sets were used as the basis for constructing the prognostic model. Subsequently, Weighted Correlation Network Analysis (WGCNA) and Cytoscape were applied to further screen the gene sets of the constructed model, and finally COX7A1 was selected for the exploration and prediction of the relationship between the clinical efficacy of immunotherapy for GC. The correlation between COX7A1 and immune cell infiltration, drug sensitivity scoring, and immunohistochemical staining were performed to initially understand the potential role of COX7A1 in the development and progression of GC. Finally, the differential expression of COX7A1 was verified in those GC patients receiving immunotherapy. RESULTS First, 47 protective genes and 408 risky genes were obtained, and the ssGSEA algorithm was applied for model construction, showing good prognostic discrimination ability. In addition, the patients with high model scores showed higher TMB and MSI levels, and lower tumor heterogeneity scores. Then, it is found that the COX7A1 expressions in GC tissues were significantly lower than those in their corresponding paracancerous tissues. Meanwhile, the patients with high COX7A1 expression showed higher probability of cancer invasion, worse clinical efficacy of immunotherapy, worse overall survival (OS) and worse disease-free survival (DFS). CONCLUSIONS The ssGSEA score we constructed can serve as a biomarker for GC patients and provide important guidance for individualized treatment. In addition, the COX7A1 gene can accurately distinguish the prognosis of GC patients and predict the clinical efficacy of immunotherapy for GC patients.
Collapse
Affiliation(s)
- Si-Yu Wang
- Department of Oncology, The First People's Hospital of Yibin, No. 65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Yu-Xin Wang
- The First Hospital of Jilin University, Changchun, 130000, China
| | - Ao Shen
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xian-Qi Yang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Cheng-Cai Liang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Run-Jie Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Rui Jian
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Nan An
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yu-Long Xiao
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Li-Shuai Wang
- Department of Oncology, The First People's Hospital of Yibin, No. 65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Yin Zhao
- Department of Oncology, The First People's Hospital of Yibin, No. 65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Chuan Lin
- Department of Oncology, The First People's Hospital of Yibin, No. 65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Chang-Ping Wang
- Department of Oncology, The First People's Hospital of Yibin, No. 65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Zhi-Ping Yuan
- Department of Oncology, The First People's Hospital of Yibin, No. 65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Shu-Qiang Yuan
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.
| |
Collapse
|
4
|
Hou L, Xing N, Yue Z, Wu J, Wang F, Guo Z. Dicer induced reactive oxygen species inhibit hepatocellular carcinoma through interacting with cytochrome c oxidase. BIOTECHNOL BIOTEC EQ 2022. [DOI: 10.1080/13102818.2022.2082318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Lin Hou
- Department of Endocrine and Metabolic Disease, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, P. R. China
| | - Na Xing
- Department of Endocrine and Metabolic Disease, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, P. R. China
| | - Zhao Yue
- Department of Gastroenterology and Hepatology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, P. R. China
| | - Jianhua Wu
- Animal Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, P. R. China
| | - Fujun Wang
- Department of Endocrine and Metabolic Disease, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, P. R. China
| | - Zhanjun Guo
- Department of Immunology and Rheumatology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, P. R. China
| |
Collapse
|
5
|
Griguer CE, Oliva CR, Coffey CS, Cudkowicz ME, Conwit RA, Gudjonsdottir AL, Ecklund DJ, Fedler JK, Neill-Hudson TM, Nabors LB, Benge M, Hackney JR, Chase M, Leonard TP, Patel T, Colman H, de la Fuente M, Chaudhary R, Marder K, Kreisl T, Mohile N, Chheda MG, McNeill K, Kumthekar P, Dogan A, Drappatz J, Puduvalli V, Kowalska A, Graber J, Gerstner E, Clark S, Salacz M, Markert J. Prospective biomarker study in newly diagnosed glioblastoma: Cyto-C clinical trial. Neurooncol Adv 2021; 4:vdab186. [PMID: 35088051 PMCID: PMC8788017 DOI: 10.1093/noajnl/vdab186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background Glioblastoma (GBM) has a 5-year survival rate of 3%-5%. GBM treatment includes maximal resection followed by radiotherapy with concomitant and adjuvant temozolomide (TMZ). Cytochrome C oxidase (CcO) is a mitochondrial enzyme involved in the mechanism of resistance to TMZ. In a prior retrospective trial, CcO activity in GBMs inversely correlated with clinical outcome. The current Cyto-C study was designed to prospectively evaluate and validate the prognostic value of tumor CcO activity in patients with newly diagnosed primary GBM, and compared to the known prognostic value of MGMT promoter methylation status. Methods This multi-institutional, blinded, prospective biomarker study enrolled 152 patients with newly diagnosed GBM who were to undergo surgical resection and would be candidates for standard of care. The primary end point was overall survival (OS) time, and the secondary end point was progression-free survival (PFS) time. Tumor CcO activity and MGMT promoter methylation status were assayed in a centralized laboratory. Results OS and PFS did not differ by high or low tumor CcO activity, and the prognostic validity of MGMT promoter methylation was confirmed. Notably, a planned exploratory analysis suggested that the combination of low CcO activity and MGMT promoter methylation in tumors may be predictive of long-term survival. Conclusions Tumor CcO activity alone was not confirmed as a prognostic marker in GBM patients. However, the combination of low CcO activity and methylated MGMT promoter may reveal a subgroup of GBM patients with improved long-term survival that warrants further evaluation. Our work also demonstrates the importance of performing large, multi-institutional, prospective studies to validate biomarkers. We also discuss lessons learned in assembling such studies.
Collapse
Affiliation(s)
- Corinne E Griguer
- Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Claudia R Oliva
- Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| | | | - Merit E Cudkowicz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robin A Conwit
- NINDS, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Dixie J Ecklund
- Department of Biostatistics, University of Iowa, Iowa City, Iowa, USA
| | - Janel K Fedler
- Department of Biostatistics, University of Iowa, Iowa City, Iowa, USA
| | | | - Louis B Nabors
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Melanie Benge
- Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James R Hackney
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Marianne Chase
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Timothy P Leonard
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Toral Patel
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Howard Colman
- Department of Neurosurgery, University of Utah, Salt Lake City, Utah, USA
| | | | - Rekha Chaudhary
- Department Internal Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Karen Marder
- Division of Neuro-Oncology, Columbia University Health Sciences, New York, New York, USA
| | - Teri Kreisl
- Division of Neuro-Oncology, Columbia University Health Sciences, New York, New York, USA
| | - Nimish Mohile
- Department of Pathology, Division of Neuropathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Milan G Chheda
- Departments of Medicine and Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Priya Kumthekar
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Aclan Dogan
- Department of Neurosurgery, Oregon Health and Science University, Portland, Oregon, USA
| | - Jan Drappatz
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Vinay Puduvalli
- Department of Neuro-Oncology, Ohio State University, Columbus, Ohio, USA
| | - Agnes Kowalska
- Department of Neurology, State University of New York, Stony Brook, New York, New York, USA
| | - Jerome Graber
- Alvord Brain Tumor Center, Swedish Medical Center, Seattle, Washington, USA
| | - Elizabeth Gerstner
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen Clark
- Department of Neurology, Vanderbilt University, Nashville, Tennessee, USA
| | - Michael Salacz
- Department Internal Medicine, University of Kansas Hospital, Kansas City, Kansas, USA
| | - James Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
6
|
RNA seq and quantitative proteomic analysis of Dictyostelium knock-out cells lacking the core autophagy proteins ATG9 and/or ATG16. BMC Genomics 2021; 22:444. [PMID: 34126926 PMCID: PMC8204557 DOI: 10.1186/s12864-021-07756-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/26/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Autophagy is an evolutionary ancient mechanism that sequesters substrates for degradation within autolysosomes. The process is driven by many autophagy-related (ATG) proteins, including the core members ATG9 and ATG16. However, the functions of these two core ATG proteins still need further elucidation. Here, we applied RNAseq and tandem mass tag (TMT) proteomic approaches to identify differentially expressed genes (DEGs) and proteins (DEPs) in Dictyostelium discoideum ATG9‾, ATG16‾ and ATG9‾/16‾ strains in comparison to AX2 wild-type cells. RESULT In total, we identified 332 (279 up and 53 down), 639 (487 up and 152 down) and 260 (114 up and 146 down) DEGs and 124 (83 up and 41 down), 431 (238 up and 193 down) and 677 (347 up and 330 down) DEPs in ATG9‾, ATG16‾ and ATG9‾/16‾ strains, respectively. Thus, in the single knock-out strains, the number of DEGs was higher than the number of DEPs while in the double knock-out strain the number of DEPs was higher. Comparison of RNAseq and proteomic data further revealed, that only a small proportion of the transcriptional changes were reflected on the protein level. Gene ontology (GO) analysis revealed an enrichment of DEPs involved in lipid metabolism and oxidative phosphorylation. Furthermore, we found increased expression of the anti-oxidant enzymes glutathione reductase (gsr) and catalase A (catA) in ATG16‾ and ATG9‾/16‾ cells, respectively, indicating adaptation to excess reactive oxygen species (ROS). CONCLUSIONS Our study provides the first combined transcriptome and proteome analysis of ATG9‾, ATG16‾ and ATG9‾/16‾ cells. Our results suggest, that most changes in protein abundance were not caused by transcriptional changes, but were rather due to changes in protein homeostasis. In particular, knock-out of atg9 and/or atg16 appears to cause dysregulation of lipid metabolism and oxidative phosphorylation.
Collapse
|
7
|
Cytochrome C Oxidase Subunit 4 (COX4): A Potential Therapeutic Target for the Treatment of Medullary Thyroid Cancer. Cancers (Basel) 2020; 12:cancers12092548. [PMID: 32911610 PMCID: PMC7565757 DOI: 10.3390/cancers12092548] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/30/2020] [Accepted: 09/02/2020] [Indexed: 12/30/2022] Open
Abstract
The nuclear-encoded subunit 4 of cytochrome c oxidase (COX4) plays a role in regulation of oxidative phosphorylation and contributes to cancer progression. We sought to determine the role of COX4 in differentiated (DTC) and medullary (MTC) thyroid cancers. We examined the expression of COX4 in human thyroid tumors by immunostaining and used shRNA-mediated knockdown of COX4 to evaluate its functional contributions in thyroid cancer cell lines. In human thyroid tissue, the expression of COX4 was higher in cancers than in either normal thyroid (p = 0.0001) or adenomas (p = 0.001). The level of COX4 expression correlated with tumor size (p = 0.04) and lymph-node metastases (p = 0.024) in patients with MTCs. COX4 silencing had no effects on cell signaling activation and mitochondrial respiration in DTC cell lines (FTC133 and BCPAP). In MTC-derived TT cells, COX4 silencing inhibited p70S6K/pS6 and p-ERK signaling, and was associated with decreased oxygen consumption and ATP production. Treatment with potassium cyanide had minimal effects on FTC133 and BCPAP, but inhibited mitochondrial respiration and induced apoptosis in MTC-derived TT cells. Our data demonstrated that metastatic MTCs are characterized by increased expression of COX4, and MTC-derived TT cells are vulnerable to COX4 silencing. These data suggest that COX4 can be considered as a novel molecular target for the treatment of MTC.
Collapse
|
8
|
Zhao L, Chen X, Feng Y, Wang G, Nawaz I, Hu L, Liu P. COX7A1 suppresses the viability of human non-small cell lung cancer cells via regulating autophagy. Cancer Med 2019; 8:7762-7773. [PMID: 31663688 PMCID: PMC6912042 DOI: 10.1002/cam4.2659] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/02/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022] Open
Abstract
COX7A1 is a subunit of cytochrome c oxidase, and plays an important role in the super‐assembly that integrates peripherally into multi‐unit heteromeric complexes in the mitochondrial respiratory chain. In recent years, some researchers have identified that COX7A1 is implicated in human cancer cell metabolism and therapy. In this study, we mainly explored the effect of COX7A1 on the cell viability of lung cancer cells. COX7A1 overexpression was induced by vector transfection in NCI‐H838 cells. Cell proliferation, colony formation and cell apoptosis were evaluated in different groups. In addition, autophagy was analyzed by detecting the expression level of p62 and LC3, as well as the tandem mRFP‐GFP‐LC3 reporter assay respectively. Our results indicated that the overexpression of COX7A1 suppressed cell proliferation and colony formation ability, and promoted cell apoptosis in human non‐small cell lung cancer cells. Besides, the overexpression of COX7A1 blocked autophagic flux and resulted in the accumulation of autophagosome via downregulation of PGC‐1α and upregulation of NOX2. Further analysis showed that the effect of COX7A1 overexpression on cell viability was partly dependent of the inhibition of autophagy. Herein, we identified that COX7A1 holds a key position in regulating the development and progression of lung cancer by affecting autophagy. Although the crosstalk among COX7A1, PGC‐1α and NOX2 needs further investigation, our study provides a novel insight into the therapeutic action of COX7A1 against human non‐small cell lung cancer.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Anesthesiology, The 2nd Clinical Medical College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, China.,Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| | - Xin Chen
- Department of Laboratory Medicine, The 2nd Clinical Medicine College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, China
| | - Yetong Feng
- Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Guangsuo Wang
- Department of Thoracic Surgery, The 2nd Clinical Medicine College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, China
| | - Imran Nawaz
- Department of Thoracic Surgery, The 2nd Clinical Medicine College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, China.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Lifu Hu
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Pengfei Liu
- Department of Anesthesiology, The 2nd Clinical Medical College (Shenzhen People's Hospital) of Jinan University, The 1st Affiliated Hospitals of Southern University of Science and Technology, Shenzhen, China.,Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| |
Collapse
|
9
|
Stein J, Tenbrock J, Kristiansen G, Müller SC, Ellinger J. Systematic expression analysis of the mitochondrial respiratory chain protein subunits identifies COX5B as a prognostic marker in clear cell renal cell carcinoma. Int J Urol 2019; 26:910-916. [PMID: 31280487 DOI: 10.1111/iju.14040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 05/19/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To analyze the expression of mitochondrial respiratory chain protein subunits in clear cell renal cell carcinoma. METHODS Possible prognostic candidates were determined using The Cancer Genome Atlas database (n = 605). The database provided valid messenger ribonucleic acid expression data for 93 genes encoding for the subunits. Selected subunits were further investigated at the messenger ribonucleic acid and protein level by real-time polymerase chain reaction, western blot and immunohistochemistry with the cohorts of the University Hospital Bonn. RESULTS The Cancer Genome Atlas messenger ribonucleic acid expression data indicated univariate and multivariate prognostic impact for seven subunits (NDUFS8, NDUFS7, COX5B, COX6B1, SDHD, COX15 and COX19). Using real-time polymerase chain reaction, significant downregulation (P < 0.05, n = 74) could be shown for COX5B, COX6B1, NDUFS7 and NDUF8 in clear cell renal cell carcinoma tissue. Survival analysis of polymerase chain reaction data showed a non-significant relationship (P = 0.067) of high COX5B expression and poor overall survival. Western blot (n = 8) and immunohistochemistry analysis (n = 167) confirmed significant COX5B downregulation on the protein level. Immunohistochemistry analysis identified COX5B as a prognostic marker for overall (P = 0.017) and cancer-specific survival (P = 0.042). CONCLUSIONS The present study findings suggest downregulation of additional subunits of mitochondrial respiratory chain proteins in clear cell renal cell carcinoma. Remarkably, COX5B, a subunit of the respiratory chain complex IV, can be identified as a novel prognostic marker.
Collapse
Affiliation(s)
- Johannes Stein
- Department of Urology, University Hospital Bonn, Bonn, Germany
| | - Julia Tenbrock
- Department of Urology, University Hospital Bonn, Bonn, Germany
| | | | - Stefan C Müller
- Department of Urology, University Hospital Bonn, Bonn, Germany
| | - Jörg Ellinger
- Department of Urology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
10
|
Oxidative Stress Induced by the Deubiquitinase Inhibitor b-AP15 Is Associated with Mitochondrial Impairment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1659468. [PMID: 31281566 PMCID: PMC6590552 DOI: 10.1155/2019/1659468] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/05/2019] [Accepted: 04/30/2019] [Indexed: 12/17/2022]
Abstract
Inhibitors of the 20S proteasome such as bortezomib are cytotoxic to tumor cells and have been proven to be valuable for the clinical management of multiple myeloma. The therapeutic efficacy of bortezomib is, however, hampered by the emergence of acquired resistance. Available data suggest that blocking proteasome activity at the level of proteasome-associated deubiquitinases (DUBs) provides a mechanism to overcome resistance to bortezomib and also to other cancer therapies. The small molecule b-AP15 is an inhibitor of proteasome-associated DUB activity that induces both proteotoxic stress and increases in the levels of reactive oxygen species (ROS) in tumor cells. Antioxidants have been shown to decrease apoptosis induction by b-AP15 and we here addressed the question of the mechanism of redox perturbation by this compound. We show that oxidative stress induction by b-AP15 is abrogated in cells deprived of mitochondrial DNA (ρ0 cells). We also show associations between the level of proteotoxic stress, the degree of mitochondrial dysfunction, and the extent of induction of hemeoxygenase-1 (HO-1), a target of the redox-regulated Nrf-2 transcription factor. Decreased expression of COX5b (cytochrome c oxidase subunit 5b) and TOMM34 (translocase of outer mitochondrial membrane 34) was observed in b-AP15-treated cells. These findings suggest a mitochondrial origin of the increased levels of ROS observed in cells exposed to the DUB inhibitor b-AP15.
Collapse
|
11
|
Wang T, Xie X, Liu H, Chen F, Du J, Wang X, Jiang X, Yu F, Fan H. Pyridine nucleotide-disulphide oxidoreductase domain 2 (PYROXD2): Role in mitochondrial function. Mitochondrion 2019; 47:114-124. [PMID: 31170524 DOI: 10.1016/j.mito.2019.05.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/23/2019] [Accepted: 05/30/2019] [Indexed: 02/05/2023]
Abstract
Pyridine Nucleotide-Disulphide Oxidoreductase Domain 2 (PYROXD2), a Hepatitis B virus X protein (HBx)-interacting protein, is significantly down-regulated in hepatocellular carcinoma (HCC), however its exact biological function remains unclear. The aim of this study is to investigate the subcellular localization and biological function of PYROXD2 in hepatic cells. The results showed that PYROXD2 was imported to the mitochondrial inner membrane/matrix by Tom40 and Tim23, but not Mia40. PYROXD2 151-230aa might be the mitochondrial targeting sequence. PYROXD2 interacted with complex IV subunit COX5B. Knockout of PYROXD2 decreased MMP, intracellular ROS, complex IV activity, cell proliferation, ATP content and mtDNA copy number, but increased mtROS levels and the number of immature mitochondria. In summary, our data illustrated that PYROXD2 localizes to the mitochondrial inner membrane/matrix, and it plays important roles in regulating mitochondrial function.
Collapse
Affiliation(s)
- Tao Wang
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - Xiaoyuan Xie
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - HuiLin Liu
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Feng Chen
- Blood center of Zhejiang province, Hangzhou, Zhejiang 310052, China
| | - Jianhua Du
- Nanchang Institute of Science and Technology, Nanchang 330108, China
| | - XingZhi Wang
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - XingYan Jiang
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - Fang Yu
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - Handong Fan
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China.
| |
Collapse
|
12
|
Zhang Y, Wang L, Lv Y, Jiang C, Wu G, Dull RO, Minshall RD, Malik AB, Hu G. The GTPase Rab1 Is Required for NLRP3 Inflammasome Activation and Inflammatory Lung Injury. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:194-206. [PMID: 30455398 PMCID: PMC6345506 DOI: 10.4049/jimmunol.1800777] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/23/2018] [Indexed: 12/22/2022]
Abstract
Uncontrolled inflammatory response during sepsis predominantly contributes to the development of multiorgan failure and lethality. However, the cellular and molecular mechanisms for excessive production and release of proinflammatory cytokines are not clearly defined. In this study, we show the crucial role of the GTPase Ras-related protein in brain (Rab)1a in regulating the nucleotide binding domain-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome activation and lung inflammatory injury. Expression of dominant negative Rab1 N124I plasmid in bone marrow-derived macrophages prevented the release of IL-1β and IL-18, NLRP3 inflammasome activation, production of pro-IL-1β and pro-IL-18, and attenuated TLR4 surface expression and NF-кB activation induced by bacterial LPS and ATP compared with control cells. In alveolar macrophage-depleted mice challenged with cecal ligation and puncture, pulmonary transplantation of Rab1a-inactivated macrophages by expression of Rab1 N124I plasmid dramatically reduced the release of IL-1β and IL-18, neutrophil count in bronchoalveolar lavage fluid, and inflammatory lung injury. Rab1a activity was elevated in alveolar macrophages from septic patients and positively associated with severity of sepsis and respiratory dysfunction. Thus, inhibition of Rab1a activity in macrophages resulting in the suppression of NLRP3 inflammasome activation may be a promising target for the treatment of patients with sepsis.
Collapse
Affiliation(s)
- Yuehui Zhang
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612
- Department of Critical Care Medicine, Affiliated Bao'an Hospital of Shenzhen, Southern Medical University, Shenzhen, Guangdong 518101, China
| | - Lijun Wang
- Department of Critical Care Medicine, Affiliated Bao'an Hospital of Shenzhen, Southern Medical University, Shenzhen, Guangdong 518101, China
| | - Yang Lv
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Chunling Jiang
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Randal O Dull
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Richard D Minshall
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612; and
| | - Asrar B Malik
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612; and
| | - Guochang Hu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612;
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612; and
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221008, China
| |
Collapse
|
13
|
Zhao H, Zhang C, Wang Y, Chen W, Alvarez PJJ. Self-Damaging Aerobic Reduction of Graphene Oxide by Escherichia coli: Role of GO-Mediated Extracellular Superoxide Formation. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:12783-12791. [PMID: 30277752 DOI: 10.1021/acs.est.8b03753] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Microbial reduction of graphene oxide (GO) under aerobic conditions is poorly understood despite its critical role in changing GO toxicity and environmental fate. Here we show that 20 mg/L GO interacts with the membrane-bound cytochrome c of E. coli in saline, shuttling electrons from the respiratory chain to extracellular molecular oxygen. This results in the formation of superoxide anions (O2•-), which in turn reduce GO in 30 min. The critical role of superoxide was demonstrated by impeding GO reduction upon addition of superoxide dismutase, or by carrying out experiments under strictly anaerobic conditions that preclude O2•- formation. Coating GO with bovine serum albumin also stopped GO reduction, which indicates the need for direct contact between GO and the cell membrane. Cell death was observed as a consequence of GO bioreduction. Apparently, electron shuttling by GO (via membrane contact) interrupts the respiratory chain and induces oxidative stress, as indicated by a 20% decrease in electron transport activity and an increase in intracellular reactive oxygen species. This novel antimicrobial mechanism could be relevant to assess GO stability and biocompatibility, and informs potential applications for microbial control.
Collapse
Affiliation(s)
- Huiru Zhao
- College of Environmental Science and Engineering , Nankai University , Tianjin 300350 , China
| | - Chengdong Zhang
- School of Environment , Beijing Normal University , Beijing 100857 , China
| | - Yaqi Wang
- College of Environmental Science and Engineering , Nankai University , Tianjin 300350 , China
| | - Wei Chen
- College of Environmental Science and Engineering , Nankai University , Tianjin 300350 , China
| | - Pedro J J Alvarez
- Department of Civil and Environmental Engineering , Rice University , Houston , Texas 77005 , United States
| |
Collapse
|
14
|
Oliveira MF, Geihs MA, França TFA, Moreira DC, Hermes-Lima M. Is "Preparation for Oxidative Stress" a Case of Physiological Conditioning Hormesis? Front Physiol 2018; 9:945. [PMID: 30116197 PMCID: PMC6082956 DOI: 10.3389/fphys.2018.00945] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/28/2018] [Indexed: 01/01/2023] Open
Affiliation(s)
- Marcus F Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcio A Geihs
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande, Brazil
| | - Thiago F A França
- Programa de Pós-Graduação em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal do Rio Grande, Rio Grande, Brazil
| | - Daniel C Moreira
- Área de Morfologia, Faculdade de Medicina, Universidade de Brasília, Brasilia, Brazil.,Departamento de Biologia Celular, Instituto de Ciências Biológicas, Universidade de Brasília, Brasilia, Brazil
| | - Marcelo Hermes-Lima
- Departamento de Biologia Celular, Instituto de Ciências Biológicas, Universidade de Brasília, Brasilia, Brazil
| |
Collapse
|
15
|
Oliva CR, Zhang W, Langford C, Suto MJ, Griguer CE. Repositioning chlorpromazine for treating chemoresistant glioma through the inhibition of cytochrome c oxidase bearing the COX4-1 regulatory subunit. Oncotarget 2018; 8:37568-37583. [PMID: 28455961 PMCID: PMC5514931 DOI: 10.18632/oncotarget.17247] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 04/07/2017] [Indexed: 12/12/2022] Open
Abstract
Patients with glioblastoma have one of the lowest overall survival rates among patients with cancer. Standard of care for patients with glioblastoma includes temozolomide and radiation therapy, yet 30% of patients do not respond to these treatments and nearly all glioblastoma tumors become resistant. Chlorpromazine is a United States Food and Drug Administration-approved phenothiazine widely used as a psychotropic in clinical practice. Recently, experimental evidence revealed the anti-proliferative activity of chlorpromazine against colon and brain tumors. Here, we used chemoresistant patient-derived glioma stem cells and chemoresistant human glioma cell lines to investigate the effects of chlorpromazine against chemoresistant glioma. Chlorpromazine selectively and significantly inhibited proliferation in chemoresistant glioma cells and glioma stem cells. Mechanistically, chlorpromazine inhibited cytochrome c oxidase (CcO, complex IV) activity from chemoresistant but not chemosensitive cells, without affecting other mitochondrial complexes. Notably, our previous studies revealed that the switch to chemoresistance in glioma cells is accompanied by a switch from the expression of CcO subunit 4 isoform 2 (COX4-2) to COX4-1. In this study, chlorpromazine induced cell cycle arrest selectively in glioma cells expressing COX4-1, and computer-simulated docking studies indicated that chlorpromazine binds more tightly to CcO expressing COX4-1 than to CcO expressing COX4-2. In orthotopic mouse brain tumor models, chlorpromazine treatment significantly increased the median overall survival of mice harboring chemoresistant tumors. These data indicate that chlorpromazine selectively inhibits the growth and proliferation of chemoresistant glioma cells expressing COX4-1. The feasibility of repositioning chlorpromazine for selectively treating chemoresistant glioma tumors should be further explored.
Collapse
Affiliation(s)
- Claudia R Oliva
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, 35294 Alabama, USA
| | - Wei Zhang
- Southern Research, Birmingham, 35294 Alabama, USA
| | - Cathy Langford
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, 35294 Alabama, USA
| | - Mark J Suto
- Southern Research, Birmingham, 35294 Alabama, USA
| | - Corinne E Griguer
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, 35294 Alabama, USA.,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, 35294 Alabama, USA
| |
Collapse
|
16
|
Garcia J, Decker CW, Sanchez SJ, Ouk JM, Siu KM, Han D. Obesity and steatosis promotes mitochondrial remodeling that enhances respiratory capacity in the liver of ob/ob mice. FEBS Lett 2018; 592:916-927. [DOI: 10.1002/1873-3468.13005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Jerome Garcia
- Department of Biology; University of La Verne; CA USA
| | - Carl W. Decker
- Department of Biopharmaceutical Sciences; School of Pharmacy; Keck Graduate Institute; Claremont CA USA
| | | | | | - Krysta M. Siu
- Department of Biology; University of La Verne; CA USA
| | - Derick Han
- Department of Biopharmaceutical Sciences; School of Pharmacy; Keck Graduate Institute; Claremont CA USA
| |
Collapse
|
17
|
West MD, Labat I, Sternberg H, Larocca D, Nasonkin I, Chapman KB, Singh R, Makarev E, Aliper A, Kazennov A, Alekseenko A, Shuvalov N, Cheskidova E, Alekseev A, Artemov A, Putin E, Mamoshina P, Pryanichnikov N, Larocca J, Copeland K, Izumchenko E, Korzinkin M, Zhavoronkov A. Use of deep neural network ensembles to identify embryonic-fetal transition markers: repression of COX7A1 in embryonic and cancer cells. Oncotarget 2017; 9:7796-7811. [PMID: 29487692 PMCID: PMC5814259 DOI: 10.18632/oncotarget.23748] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/20/2017] [Indexed: 12/19/2022] Open
Abstract
Here we present the application of deep neural network (DNN) ensembles trained on transcriptomic data to identify the novel markers associated with the mammalian embryonic-fetal transition (EFT). Molecular markers of this process could provide important insights into regulatory mechanisms of normal development, epimorphic tissue regeneration and cancer. Subsequent analysis of the most significant genes behind the DNNs classifier on an independent dataset of adult-derived and human embryonic stem cell (hESC)-derived progenitor cell lines led to the identification of COX7A1 gene as a potential EFT marker. COX7A1, encoding a cytochrome C oxidase subunit, was up-regulated in post-EFT murine and human cells including adult stem cells, but was not expressed in pre-EFT pluripotent embryonic stem cells or their in vitro-derived progeny. COX7A1 expression level was observed to be undetectable or low in multiple sarcoma and carcinoma cell lines as compared to normal controls. The knockout of the gene in mice led to a marked glycolytic shift reminiscent of the Warburg effect that occurs in cancer cells. The DNN approach facilitated the elucidation of a potentially new biomarker of cancer and pre-EFT cells, the embryo-onco phenotype, which may potentially be used as a target for controlling the embryonic-fetal transition.
Collapse
Affiliation(s)
| | - Ivan Labat
- AgeX Therapeutics, Inc., Alameda, CA, USA
| | | | | | | | | | | | - Eugene Makarev
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA
| | - Alex Aliper
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA
| | - Andrey Kazennov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Andrey Alekseenko
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,Innopolis University, Innoplis, Russia
| | - Nikolai Shuvalov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Evgenia Cheskidova
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Aleksandr Alekseev
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Artem Artemov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA
| | - Evgeny Putin
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,Computer Technologies Lab, ITMO University, St. Petersburg, Russia
| | - Polina Mamoshina
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA
| | - Nikita Pryanichnikov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA
| | | | | | - Evgeny Izumchenko
- Johns Hopkins University, School of Medicine, Department of Otolaryngology-Head and Neck Cancer Research, Baltimore, MD, USA
| | - Mikhail Korzinkin
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA
| | - Alex Zhavoronkov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University at Eastern, Baltimore, MD, USA.,The Biogerontology Research Foundation, Trevissome Park, Truro, UK
| |
Collapse
|
18
|
Lau GY, Mandic M, Richards JG. Evolution of Cytochrome c Oxidase in Hypoxia Tolerant Sculpins (Cottidae, Actinopterygii). Mol Biol Evol 2017; 34:2153-2162. [PMID: 28655155 DOI: 10.1093/molbev/msx179] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Vertebrate hypoxia tolerance can emerge from modifications to the oxygen (O2) transport cascade, but whether there is adaptive variation to O2 binding at the terminus of this cascade, mitochondrial cytochrome c oxidase (COX), is not known. In order to address the hypothesis that hypoxia tolerance is associated with enhanced O2 binding by mitochondria we undertook a comparative analysis of COX O2 kinetics across species of intertidal sculpins (Cottidae, Actinopterygii) that vary in hypoxia tolerance. Our analysis revealed a significant relationship between hypoxia tolerance (critical O2 tension of O2 consumption rate; Pcrit), mitochondrial O2 binding affinity (O2 tension at which mitochondrial respiration was half maximal; P50), and COX O2-binding affinity (apparent Michaelis-Menten constant for O2 binding to COX; Km,app O2). The more hypoxia tolerant species had both a lower mitochondrial P50 and lower COX Km,app O2, facilitating the maintenance of mitochondrial function to a lower O2 tension than in hypoxia intolerant species. Additionally, hypoxia tolerant species had a lower overall COX Vmax but higher mitochondrial COX respiration rate when expressed relative to maximal electron transport system respiration rate. In silico analyses of the COX3 subunit postulated as the entry point for O2 into the COX protein catalytic core, points to variation in COX3 protein stability (estimated as free energy of unfolding) contributing to the variation in COX Km,app O2. We propose that interactions between COX3 and cardiolipin at four amino acid positions along the same alpha-helix forming the COX3 v-cleft represent likely determinants of interspecific differences in COX Km,app O2.
Collapse
Affiliation(s)
- Gigi Y Lau
- Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| | - Milica Mandic
- Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| | - Jeffrey G Richards
- Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
19
|
Mishra N, Timilsina U, Ghimire D, Dubey RC, Gaur R. Downregulation of cytochrome c oxidase subunit 7A1 expression is important in enhancing cell proliferation in adenocarcinoma cells. Biochem Biophys Res Commun 2017; 482:713-719. [DOI: 10.1016/j.bbrc.2016.11.100] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 11/17/2016] [Indexed: 10/20/2022]
|
20
|
Chen W, Xia X, Song N, Wang Y, Zhu H, Deng W, Kong Q, Pan X, Qin C. Cross-Species Analysis of Gene Expression and Function in Prefrontal Cortex, Hippocampus and Striatum. PLoS One 2016; 11:e0164295. [PMID: 27716781 PMCID: PMC5055290 DOI: 10.1371/journal.pone.0164295] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 09/22/2016] [Indexed: 01/08/2023] Open
Abstract
Background Mouse has been extensively used as a tool for investigating the onset and development of human neurological disorders. As a first step to construct a transgenic mouse model of human brain lesions, it is of fundamental importance to clarify the similarity and divergence of genetic background between non-diseased human and mouse brain tissues. Methods We systematically compared, based on large scale integrated microarray data, the transcriptomes of three anatomically distinct brain regions; prefrontal cortex (PFC), hippocampus (HIP) and striatum (STR), across human and mouse. The widely used DAVID web server was used to decipher the biological functions of the highly expressed genes that were identified using a previously reported approach. Venn analysis was used to depict the overlapping ratios of the notably enriched biological process (BP) terms (one-tailed Fisher’s exact test and Benjamini correction; adjusted p < 0.01) between two brain tissues. GOSemSim, an R package, was selected to perform GO semantic similarity analysis. Next, we adjusted signal intensities of orthologous genes by the total signals in all samples within species, and used one minus Pearson’s correlation coefficient to assess the expression distance. Hierarchical clustering and principal component analysis (PCA) were selected for expression pattern analysis. Lineage specific expressed orthologous genes were identified by comparison of the most extreme sub-datasets across species and further verified using reverse transcription PCR (RT-PCR) and quantitative real-time PCR (qRT-PCR). Results We found that the number of the significantly enriched BP terms of the highly expressed genes in human brain regions is larger than that in mouse corresponding brain regions. The mainly involved BP terms in human brain tissues associated with protein-membrane targeting and selenium metabolism are species-specific. The overlapping ratios of all the significantly enriched BP terms between any two brain tissues across species are lower than that within species, but the pairwise semantic similarities are very high between any two brain tissues from either human or mouse. Hierarchical clustering analysis shows the biological functions of the highly expressed genes in brain tissues are more consistent within species than interspecies; whereas it shows the expression patterns of orthologous genes are evidently conserved between human and mouse equivalent brain tissues. In addition, we identified four orthologous genes (COX5B, WIF1, SLC4A10 and PLA2G7) that are species-specific, which have been widely studied and confirmed to be closely linked with neuro- physiological and pathological functions. Conclusion Our study highlights the similarities and divergences in gene function and expression between human and mouse corresponding brain regions, including PFC, HIP and STR.
Collapse
Affiliation(s)
- Wei Chen
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P.R. China
| | - Xiayu Xia
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P.R. China
| | - Nan Song
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P.R. China
| | - Ying Wang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P.R. China
| | - Hua Zhu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P.R. China
| | - Wei Deng
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P.R. China
| | - Qi Kong
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P.R. China
| | - Xianmin Pan
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P.R. China
- Ministry of Education, The Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, P.R. China
| | - Chuan Qin
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P.R. China
- * E-mail:
| |
Collapse
|
21
|
Huang YP, Chang NW. PPARα modulates gene expression profiles of mitochondrial energy metabolism in oral tumorigenesis. Biomedicine (Taipei) 2016; 6:3. [PMID: 26869356 PMCID: PMC4751096 DOI: 10.7603/s40681-016-0003-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 08/15/2015] [Indexed: 11/16/2022] Open
Abstract
Metabolic reprogramming plays a crucial role in the development of cancer. The aim of this study was to explore the effect of fenofibrate, an agonist of peroxisome proliferator-activated receptor alpha (PPARα), on gene expression profiles of mitochondrial energy metabolism. Our results showed that PPARα expression was negatively correlated with tumor progression in an oral cancer mouse model. Activation of PPARα through fenofibrate suppressed migration of oral cancer cells. Differential protein profiling demonstrated that expressions of genes related to mitochondrial energy metabolism were either up-regulated (Atp5g3, Cyc1, Ndufa5, Ndufa10, and Sdhd) or down-regulated (Cox5b, Ndufa1, Ndufb7, and Uqcrh) through PPARα activation and response. Our results indicate that PPARα exhibits a great potential for anti-oral cancer therapies by modulating cancer cell mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Yi-Ping Huang
- Department of Physiology, College of Medicine, China Medical University, 404, Taichung, Taiwan
| | - Nai Wen Chang
- Department of Biochemistry, College of Medicine,, China Medical University, 404, No. 91, Hsueh-Shih Road, Taichung, Taiwan.
| |
Collapse
|
22
|
Nuclear-encoded cytochrome c oxidase subunit 4 regulates BMI1 expression and determines proliferative capacity of high-grade gliomas. Oncotarget 2015; 6:4330-44. [PMID: 25726526 PMCID: PMC4414193 DOI: 10.18632/oncotarget.3015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 12/31/2014] [Indexed: 12/20/2022] Open
Abstract
Nuclear-encoded cytochrome c oxidase subunit 4 (COX4) is a key regulatory subunit of mammalian cytochrome c oxidase, and recent studies have demonstrated that COX4 isoform 1 (COX4-1) could have a role in glioma chemoresistance. The Polycomb complex protein BMI1 is a stem cell regulatory gene implicated in the pathogenesis of many aggressive cancers, including glioma. This study sought to determine if COX4 regulates BMI1 and modulates tumor cell proliferation. Using The Cancer Genome Atlas database and a retrospective data set from patients with glioblastoma multiforme, we found that BMI1 expression levels positively correlated with COX4-1 expression and overall survival. Whereas COX4-1 promoted cell growth by increasing BMI1 expression, COX4-2 inhibited cell growth even in cells overexpressing BMI1. We also demonstrate that COX4-1 attenuates mitochondrial reactive oxygen species (ROS) production, which is required for COX4-1-mediated effects on BMI1 expression and cell proliferation. Notably, mice bearing COX4-1-expressing glioma cell xenografts quickly developed invasive tumors characterized by the presence of multiple lesions positive for Ki-67, BMI1, and COX4-1, whereas mice bearing COX4-2-expressing xenografts rarely developed tumors by this point. COX4-1 also promoted the self-renewal of glioma stem-like cells, consistent with the reported role of BMI1 in stem cell growth. Taken together, these findings identify a novel COX4-1-mitochondrial ROS axis, in which differential expression of COX4 isoforms regulates mitochondrial ROS production and controls BMI1 expression.
Collapse
|
23
|
Xing WM, Yuan TJ, Xu JD, Gu LL, Liang P, Lu H. Proteomic identification of mitochondrial targets involved in andrographolide sodium bisulfite-induced nephrotoxicity in a rat model. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 40:592-599. [PMID: 26356389 DOI: 10.1016/j.etap.2015.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 08/07/2015] [Accepted: 08/10/2015] [Indexed: 06/05/2023]
Abstract
Our previous works have indicated that the mitochondrion is the primary target of nephrotoxicity induced by andrographolide sodium bisulfate (ASB), but the mechanisms of ASB-induced nephrotoxicity have remained largely unknown. In this study, proteomic analysis was used to explore the changes in the renal mitochondrial proteome in SD rats after treatment with ASB. SD rats were intraperitoneally administered with ASB (100, 600mg/kg/d) for 7 days. Renal impairment was evaluated by pathological observation. Two-dimensional gel electrophoresis (2-DE), as well as matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry (MS), was applied for the identification of mitochondrial protein and was validated by Western blotting. Protein-protein interactions were analyzed using a Web-based bioinformatics tool (STRING, version 9.1). Rat kidneys exhibited histopathological changes after treatment with ASB, and 13 proteins were significantly changed, including ES1 protein homolog, heat shock cognate 71kDa protein, peroxiredoxin-1 (Prdx1), cytochrome C oxidase subunit 5B (COX5B), prohibitin (PHB), threonine-tRNA ligase, pyruvate dehydrogenase E1 component subunit beta (PDH-β), voltage-dependent anion-selective channel protein 2 (VDAC2), voltage-dependent anion-selective channel protein 1 (VDAC1), adenylate kinase 2 (KAD2) and others. These data demonstrated that the expression levels of several proteins significantly changed in the mitochondria, and these proteins could be candidate biomarkers for ASB-induced nephrotoxicity.
Collapse
Affiliation(s)
- Wen Min Xing
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Tang Juan Yuan
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Jia Dong Xu
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Li Li Gu
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Pei Liang
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Hong Lu
- School of Pharmacology, Zhejiang Chinese Medical University, Hangzhou, PR China.
| |
Collapse
|
24
|
Du C, Liu HF, Lin YZ, Wang XF, Ma J, Li YJ, Wang X, Zhou JH. Proteomic alteration of equine monocyte-derived macrophages infected with equine infectious anemia virus. Proteomics 2015; 15:1843-58. [PMID: 25684102 DOI: 10.1002/pmic.201400279] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 01/06/2015] [Accepted: 02/05/2015] [Indexed: 01/18/2023]
Abstract
Similar to the well-studied viruses human immunodeficiency virus (HIV)-1 and simian immunodeficiency virus (SIV), equine infectious anemia virus (EIAV) is another member of the Lentivirus genus in the family Retroviridae. Previous studies revealed that interactions between EIAV and the host resulted in viral evolution in pathogenicity and immunogenicity, as well as adaptation to the host. Proteomic analysis has been performed to examine changes in protein expression and/or modification in host cells infected with viruses and has revealed useful information for virus-host interactions. In this study, altered protein expression in equine monocyte-derived macrophages (eMDMs, the principle target cell of EIAV in vivo) infected with the EIAV pathogenic strain EIAV(DLV34) (DLV34) was examined using 2D-LC-MS/MS coupled with the iTRAQ labeling technique. The expression levels of 210 cellular proteins were identified to be significantly upregulated or downregulated by infection with DLV34. Alterations in protein expression were confirmed by examining the mRNA levels of eight selected proteins using quantitative real-time reverse-transcription PCR, and by verifying the levels of ten selected proteins using parallel reaction monitoring (PRM). Further analysis of GO and Kyoto Encyclopedia of Genes and Genomes (KEGG)-Pathway enrichment demonstrated that these differentially expressed proteins are primarily related to the biological processes of oxidative phosphorylation, protein folding, RNA splicing, and ubiquitylation. Our results can facilitate a better understanding of the host response to EIAV infection and the cellular processes required for EIAV replication and pathogenesis.
Collapse
Affiliation(s)
- Cheng Du
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, P. R. China.,Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Hai-Fang Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, P. R. China
| | - Yue-Zhi Lin
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, P. R. China
| | - Xue-Feng Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, P. R. China
| | - Jian Ma
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, P. R. China
| | - Yi-Jing Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, P. R. China
| | - Xiaojun Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, P. R. China
| | - Jian-Hua Zhou
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, P. R. China.,Hayao Pharmaceutical Group Biovaccine Co, Harbin, P. R. China
| |
Collapse
|
25
|
Klichko V, Sohal BH, Radyuk SN, Orr WC, Sohal RS. Decrease in cytochrome c oxidase reserve capacity diminishes robustness of Drosophila melanogaster and shortens lifespan. Biochem J 2014; 459:127-35. [PMID: 24444354 DOI: 10.1042/bj20131370] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The phenotypic effects of under- and over-expression of CcO (cytochrome c oxidase) regulatory subunits IV and Vb were examined in Drosophila melanogaster in order to test further the hypothesis that suppression of the activities of mitochondrial ETC (electron-transport chain) oxidoreductases retards the aging process and extends lifespan. Underexpression of both CcO subunits, induced by RNAi, resulted in decreases in the respective mRNA and protein levels, CcO holoenzyme activity, rate of mitochondrial respiration, walking speed and the lifespan of fruitflies. Overexpression of CcO IV or Vb in young fruitflies increased the amount of mRNA, but had no effect on the protein level or CcO catalytic activity. On the other hand, in older fruitflies, overexpression of CcO Vb, but not CcO IV, elevated the mRNA and protein amounts as well as the CcO holoenzyme activity, thereby preventing the typical age-related decline in CcO activity. Nevertheless, lifespans of the fruitflies overexpressing CcO IV or Vb were neither extended nor shortened. Our results demonstrate that: (i) the suppression of CcO function exerts deleterious rather than benign effects on fitness and survival, and (ii) the structure/function of CcO, an ETC oxidoreductase, can be 're-engineered' in vivo.
Collapse
Affiliation(s)
- Vladimir Klichko
- *Department of Biological Sciences, Southern Methodist University, Dallas, TX 75275, U.S.A
| | - Barbara H Sohal
- †Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, U.S.A
| | - Svetlana N Radyuk
- *Department of Biological Sciences, Southern Methodist University, Dallas, TX 75275, U.S.A
| | - William C Orr
- *Department of Biological Sciences, Southern Methodist University, Dallas, TX 75275, U.S.A
| | - Rajindar S Sohal
- †Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, U.S.A
| |
Collapse
|
26
|
Jacobs JL, Coyne CB. Mechanisms of MAVS regulation at the mitochondrial membrane. J Mol Biol 2013; 425:5009-19. [PMID: 24120683 DOI: 10.1016/j.jmb.2013.10.007] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/02/2013] [Accepted: 10/02/2013] [Indexed: 12/24/2022]
Abstract
Mitochondria have emerged as critical platforms for antiviral innate immune signaling. This is due in large part to the mitochondrial localization of the innate immune signaling adaptor MAVS (mitochondrial antiviral signaling protein), which coordinates signals received from two independent cytosolic pathogen recognition receptors (PRRs) to induce antiviral genes. The existence of a shared adaptor for two central PRRs presents an ideal target by which the host cell can prevent cellular damage induced by uncontrolled inflammation through alteration of MAVS expression and/or signaling. In this review, we focus on the MAVS regulome and review the cellular factors that regulate MAVS by (1) protein-protein interactions, (2) alterations in mitochondrial dynamics, and/or (3) post-translational modifications.
Collapse
Affiliation(s)
- Jana L Jacobs
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | | |
Collapse
|
27
|
Henson AL, Moore JB, Alard P, Wattenberg MM, Liu JM, Ellis SR. Mitochondrial function is impaired in yeast and human cellular models of Shwachman Diamond syndrome. Biochem Biophys Res Commun 2013; 437:29-34. [PMID: 23792098 DOI: 10.1016/j.bbrc.2013.06.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 06/10/2013] [Indexed: 12/22/2022]
Abstract
Shwachman Diamond syndrome (SDS) is an inherited bone marrow failure syndrome typically characterized by neutropenia, exocrine pancreas dysfunction, metaphyseal chondrodysplasia, and predisposition to myelodysplastic syndrome and leukemia. SBDS, the gene affected in most cases of SDS, encodes a protein known to influence many cellular processes including ribosome biogenesis, mitotic spindle assembly, chemotaxis, and the regulation of reactive oxygen species production. The best characterized role for the SBDS protein is in the production of functional 60S ribosomal subunits. Given that a reduction in functional 60S subunits could impact on the translational output of cells depleted of SBDS we analyzed protein synthesis in yeast cells lacking SDO1, the ortholog of SBDS. Cells lacking SDO1 selectively increased the synthesis of POR1, the ortholog of mammalian VDAC1 a major anion channel of the mitochondrial outer membrane. Further studies revealed the cells lacking SDO1 were compromised in growth on non-fermentable carbon sources suggesting mitochondrial function was impaired. These observations prompted us to examine mitochondrial function in human cells where SBDS expression was reduced. Our studies indicate that reduced expression of SBDS decreases mitochondrial membrane potential and oxygen consumption and increases the production of reactive oxygen species. These studies indicate that mitochondrial function is also perturbed in cells expressing reduced amounts of SBDS and indicate that disruption of mitochondrial function may also contribute to SDS pathophysiology.
Collapse
Affiliation(s)
- Adrianna L Henson
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY 40292, United States
| | | | | | | | | | | |
Collapse
|
28
|
Griguer CE, Cantor AB, Fathallah-Shaykh HM, Gillespie GY, Gordon AS, Markert JM, Radovanovic I, Clement-Schatlo V, Shannon CN, Oliva CR. Prognostic relevance of cytochrome C oxidase in primary glioblastoma multiforme. PLoS One 2013; 8:e61035. [PMID: 23593382 PMCID: PMC3622606 DOI: 10.1371/journal.pone.0061035] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Accepted: 03/05/2013] [Indexed: 12/13/2022] Open
Abstract
Patients with primary glioblastoma multiforme (GBM) have one of the lowest overall survival rates among cancer patients, and reliable biomarkers are necessary to predict patient outcome. Cytochrome c oxidase (CcO) promotes the switch from glycolytic to OXPHOS metabolism, and increased CcO activity in tumors has been associated with tumor progression after chemotherapy failure. Thus, we investigated the relationship between tumor CcO activity and the survival of patients diagnosed with primary GBM. A total of 84 patients with grade IV glioma were evaluated in this retrospective cohort study. Cumulative survival was calculated by the Kaplan-Meier method and analyzed by the log-rank test, and univariate and multivariate analyses were performed with the Cox regression model. Mitochondrial CcO activity was determined by spectrophotometrically measuring the oxidation of cytochrome c. High CcO activity was detected in a subset of glioma tumors (∼30%), and was an independent prognostic factor for shorter progression-free survival and overall survival [P = 0.0087 by the log-rank test, hazard ratio = 3.57 for progression-free survival; P<0.001 by the log-rank test, hazard ratio = 10.75 for overall survival]. The median survival time for patients with low tumor CcO activity was 14.3 months, compared with 6.3 months for patients with high tumor CcO activity. High CcO activity occurs in a significant subset of high-grade glioma patients and is an independent predictor of poor outcome. Thus, CcO activity may serve as a useful molecular marker for the categorization and targeted therapy of GBMs.
Collapse
Affiliation(s)
- Corinne E Griguer
- Division of Neurosurgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States of America.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Zhao Y, Sun X, Nie X, Sun L, Tang TS, Chen D, Sun Q. COX5B regulates MAVS-mediated antiviral signaling through interaction with ATG5 and repressing ROS production. PLoS Pathog 2012; 8:e1003086. [PMID: 23308066 PMCID: PMC3534373 DOI: 10.1371/journal.ppat.1003086] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 10/29/2012] [Indexed: 12/26/2022] Open
Abstract
Innate antiviral immunity is the first line of the host defense system that rapidly detects invading viruses. Mitochondria function as platforms for innate antiviral signal transduction in mammals through the adaptor protein, MAVS. Excessive activation of MAVS-mediated antiviral signaling leads to dysfunction of mitochondria and cell apoptosis that likely causes the pathogenesis of autoimmunity. However, the mechanism of how MAVS is regulated at mitochondria remains unknown. Here we show that the Cytochrome c Oxidase (CcO) complex subunit COX5B physically interacts with MAVS and negatively regulates the MAVS-mediated antiviral pathway. Mechanistically, we find that while activation of MAVS leads to increased ROS production and COX5B expression, COX5B down-regulated MAVS signaling by repressing ROS production. Importantly, our study reveals that COX5B coordinates with the autophagy pathway to control MAVS aggregation, thereby balancing the antiviral signaling activity. Thus, our study provides novel insights into the link between mitochondrial electron transport system and the autophagy pathway in regulating innate antiviral immunity. Pattern recognition receptors are vital to innate immunity. In the antiviral innate immunity, retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), such as RIG-I and MDA5, sense viral RNAs through their C-terminal helicase domains, then initiate the antiviral response through interaction with the essential adaptor protein MAVS, which is located in mitochondrial outer membrane. Although cumulative studies have showed that mitochondria-associated MAVS plays an important role in antiviral signaling, much remains unknown about the mechanism of MAVS activity related to mitochondrial membrane localization. In this article we demonstrate that the CcO complex subunit COX5B negatively regulates the MAVS-mediated antiviral pathway through interaction with MAVS. At the mechanistic level, we show that COX5B inhibits MAVS-mediated antiviral pathway by suppressing ROS production, and coordinating with the autophagy pathway to control MAVS aggregation. Our data support a notion that mitochondrial electron transport system coordinates with the autophagy pathway to regulate MAVS-mediated signaling for a tight control of innate antiviral immunity.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, P. R. China
| | - Xiaofeng Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, P. R. China
| | - Xuanli Nie
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, P. R. China
| | - Liwei Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, P. R. China
| | - Tie-shan Tang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, P. R. China
| | - Dahua Chen
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, P. R. China
| | - Qinmiao Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, P. R. China
- * E-mail:
| |
Collapse
|
30
|
Han D, Ybanez MD, Johnson HS, McDonald JN, Mesropyan L, Sancheti H, Martin G, Martin A, Lim AM, Dara L, Cadenas E, Tsukamoto H, Kaplowitz N. Dynamic adaptation of liver mitochondria to chronic alcohol feeding in mice: biogenesis, remodeling, and functional alterations. J Biol Chem 2012; 287:42165-79. [PMID: 23086958 DOI: 10.1074/jbc.m112.377374] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Liver mitochondria undergo dynamic alterations following chronic alcohol feeding to mice. Intragastric alcohol feeding to mice resulted in 1) increased state III respiration (109% compared with control) in isolated liver mitochondria, probably due to increased levels of complexes I, IV, and V being incorporated into the respiratory chain; 2) increased mitochondrial NAD(+) and NADH levels (∼2-fold), with no change in the redox status; 3) alteration in mitochondrial morphology, with increased numbers of elongated mitochondria; and 4) enhanced mitochondrial biogenesis in the liver, which corresponded with an up-regulation of PGC-1α (peroxisome proliferator-activated receptor γ coactivator-1α). Oral alcohol feeding to mice, which is associated with less liver injury and steatosis, slightly enhanced respiration in isolated liver mitochondria (30.8% compared with control), lower than the striking increase caused by intragastric alcohol feeding. Mitochondrial respiration increased with both oral and intragastric alcohol feeding despite extensive N-acetylation of mitochondrial proteins. The alcohol-induced mitochondrial alterations are probably an adaptive response to enhance alcohol metabolism in the liver. Isolated liver mitochondria from alcohol-treated mice had a greater rate of acetaldehyde metabolism and respiration when treated with acetaldehyde than control. Aldehyde dehydrogenase-2 levels were unaltered in response to alcohol, suggesting that the greater acetaldehyde metabolism by isolated mitochondria from alcohol-treated mice was due to increased mitochondrial respiration that regenerated NAD(+), the rate-limiting substrate in alcohol/acetaldehyde metabolism. Overall, our work suggests that mitochondrial plasticity in the liver may be an important adaptive response to the metabolic stress caused by alcohol intake and could potentially play a role in many other vital functions performed by the liver.
Collapse
Affiliation(s)
- Derick Han
- University of Southern California Research Center for Liver Diseases and Southern California Research Center for Alcoholic Liver and Pancreatic Diseases, Keck School of Medicine, University of Southern California, Los Angeles, California 90089-9121, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Telang S, Nelson KK, Siow DL, Yalcin A, Thornburg JM, Imbert-Fernandez Y, Klarer AC, Farghaly H, Clem BF, Eaton JW, Chesney J. Cytochrome c oxidase is activated by the oncoprotein Ras and is required for A549 lung adenocarcinoma growth. Mol Cancer 2012; 11:60. [PMID: 22917272 PMCID: PMC3546037 DOI: 10.1186/1476-4598-11-60] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 08/17/2012] [Indexed: 12/12/2022] Open
Abstract
Background Constitutive activation of Ras in immortalized bronchial epithelial cells increases electron transport chain activity, oxygen consumption and tricarboxylic acid cycling through unknown mechanisms. We hypothesized that members of the Ras family may stimulate respiration by enhancing the expression of the Vb regulatory subunit of cytochrome c oxidase (COX). Results We found that the introduction of activated H-RasV12 into immortalized human bronchial epithelial cells increased eIF4E-dependent COX Vb protein expression simultaneously with an increase in COX activity and oxygen consumption. In support of the regulation of COX Vb expression by the Ras family, we also found that selective siRNA-mediated inhibition of K-Ras expression in A549 lung adenocarcinoma cells reduced COX Vb protein expression, COX activity, oxygen consumption and the steady-state concentration of ATP. We postulated that COX Vb-mediated activation of COX activity may be required for the anchorage-independent growth of A549 cells as soft agar colonies or as lung xenografts. We transfected the A549 cells with COX Vb small interfering or shRNA and observed a significant reduction of their COX activity, oxygen consumption, ATP and ability to grow in soft agar and as poorly differentiated tumors in athymic mice. Conclusion Taken together, our findings indicate that the activation of Ras increases COX activity and mitochondrial respiration in part via up-regulation of COX Vb and that this regulatory subunit of COX may have utility as a Ras effector target for the development of anti-neoplastic agents.
Collapse
Affiliation(s)
- Sucheta Telang
- Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ali SS, Hsiao M, Zhao HW, Dugan LL, Haddad GG, Zhou D. Hypoxia-adaptation involves mitochondrial metabolic depression and decreased ROS leakage. PLoS One 2012; 7:e36801. [PMID: 22574227 PMCID: PMC3344937 DOI: 10.1371/journal.pone.0036801] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 04/13/2012] [Indexed: 11/19/2022] Open
Abstract
Through long-term laboratory selection, we have generated a Drosophila melanogaster population that tolerates severe, normally lethal, level of hypoxia. This strain lives perpetually under severe hypoxic conditions (4% O(2)). In order to shed light on the mechanisms involved in this adaptation, we studied the respiratory function of isolated mitochondria from the thorax of hypoxia-adapted flies (AF) using polarographic oxygen consumption while monitoring superoxide generation by electron paramagnetic resonance (EPR) techniques. AF mitochondria exhibited a significant 30% decrease in respiratory rate during state 3, while enhancing the resting respiratory rate during State 4-oligo by 220%. The activity of individual electron transport complexes I, II and III were 107%, 65%, and 120% in AF mitochondria as compared to those isolated from control flies. The sharp decrease in complex II activity and modest increase in complexes I and III resulted in >60% reduction in superoxide leakage from AF mitochondria during both NAD(+)-linked state 3 and State 4-oligo respirations. These results provide evidence that flies with mitochondria exhibiting decreased succinate dehydrogenase activity and reduced superoxide leakage give flies an advantage for survival in long-term hypoxia.
Collapse
Affiliation(s)
- Sameh S. Ali
- Department of Medicine, University of California San Diego, San Diego, California, United States of America
- The Center for Aging and Associated Diseases, Helmy Institute of Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- * E-mail: (SSA); (DZ)
| | - Mary Hsiao
- Department of Pediatrics, University of California San Diego, San Diego, California, United States of America
| | - Huiwen W. Zhao
- Department of Pediatrics, University of California San Diego, San Diego, California, United States of America
| | - Laura L. Dugan
- Department of Medicine, University of California San Diego, San Diego, California, United States of America
- Department of Neuroscience, University of California San Diego, San Diego, California, United States of America
| | - Gabriel G. Haddad
- Department of Pediatrics, University of California San Diego, San Diego, California, United States of America
- Department of Neuroscience, University of California San Diego, San Diego, California, United States of America
- Rady Children's Hospital-San Diego, San Diego, California, United States of America
| | - Dan Zhou
- Department of Pediatrics, University of California San Diego, San Diego, California, United States of America
- * E-mail: (SSA); (DZ)
| |
Collapse
|
33
|
Mitochondria: redox metabolism and dysfunction. Biochem Res Int 2012; 2012:896751. [PMID: 22593827 PMCID: PMC3347708 DOI: 10.1155/2012/896751] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 02/05/2012] [Indexed: 02/07/2023] Open
Abstract
Mitochondria are the main intracellular location for fuel generation; however, they are not just power plants but involved in a range of other intracellular functions including regulation of redox homeostasis and cell fate. Dysfunction of mitochondria will result in oxidative stress which is one of the underlying causal factors for a variety of diseases including neurodegenerative diseases, diabetes, cardiovascular diseases, and cancer. In this paper, generation of reactive oxygen/nitrogen species (ROS/RNS) in the mitochondria, redox regulatory roles of certain mitochondrial proteins, and the impact on cell fate will be discussed. The current state of our understanding in mitochondrial dysfunction in pathological states and how we could target them for therapeutic purpose will also be briefly reviewed.
Collapse
|
34
|
Low ICC, Kang J, Pervaiz S. Bcl-2: a prime regulator of mitochondrial redox metabolism in cancer cells. Antioxid Redox Signal 2011; 15:2975-87. [PMID: 21574773 DOI: 10.1089/ars.2010.3851] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
SIGNIFICANCE Mitochondria play a critical role as death amplifiers during drug-induced apoptosis in cancer cells by providing pro-apoptotic factors that are released from the mitochondrial inter-membranous space upon the induction of mitochondrial outer membrane permeabilization. This intrinsic death signaling pathway is the preferred mechanism employed by most anticancer compounds, and as such, resistance to drug-induced apoptosis is invariably associated with inhibition of mitochondrial death signaling network. The latter is a function of a balance between the pro- and the anti-apoptotic members of the Bcl-2 family. Bcl-2 is the prototype anti-apoptotic protein that localizes to the mitochondria and blocks the recruitment and activation of pro-apoptotic proteins, such as Bax, to the mitochondria. RECENT ADVANCES AND CRITICAL ISSUES Recent evidence has highlighted a novel mechanism of anti-apoptotic activity of Bcl-2 in addition to its canonical activity in regulating mitochondrial outer membrane permeabilization. This novel activity is a function of cellular redox regulation, in particular, mitochondrial metabolism in cancer cells. FUTURE DIRECTIONS Here we review the current state of our understanding of the death inhibitory activity of Bcl-2 and provide insight into the novel functional biology of this remarkable protein, which could have implications for designing innovative strategies to overcome the problem of drug resistance in the clinical settings.
Collapse
Affiliation(s)
- Ivan Cherh Chiet Low
- ROS, Apoptosis and Cancer Biology Laboratory, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | |
Collapse
|
35
|
Sturrock A, Seedahmed E, Mir-Kasimov M, Boltax J, McManus ML, Paine R. GM-CSF provides autocrine protection for murine alveolar epithelial cells from oxidant-induced mitochondrial injury. Am J Physiol Lung Cell Mol Physiol 2011; 302:L343-51. [PMID: 22140071 DOI: 10.1152/ajplung.00276.2011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Exposure of mice to hyperoxia induces alveolar epithelial cell (AEC) injury, acute lung injury and death. Overexpression of granulocyte-macrophage colony-stimulating factor (GM-CSF) in the lung protects against these effects, although the mechanisms are not yet clear. Hyperoxia induces cellular injury via effects on mitochondrial integrity, associated with induction of proapoptotic members of the Bcl-2 family. We hypothesized that GM-CSF protects AEC through effects on mitochondrial integrity. MLE-12 cells (a murine type II cell line) and primary murine type II AEC were subjected to oxidative stress by exposure to 80% oxygen and by exposure to H(2)O(2). Exposure to H(2)O(2) induced cytochrome c release and decreased mitochondrial reductase activity in MLE-12 cells. Incubation with GM-CSF significantly attenuated these effects. Protection induced by GM-CSF was associated with Akt activation. GM-CSF treatment also resulted in increased expression of the antiapoptotic Bcl-2 family member, Mcl-1. Primary murine AEC were significantly more tolerant of oxidative stress than MLE-12 cells. In contrast to MLE-12 cells, primary AEC expressed significant GM-CSF at baseline and demonstrated constitutive activation of Akt and increased baseline expression of Mcl-1. Treatment with exogenous GM-CSF further increased Akt activation and Mcl-1 expression in primary AEC. Conversely, suppression of AEC GM-CSF expression by use of GM-CSF-specific small interfering RNA resulted in decreased tolerance of oxidative stress, Furthermore, silencing of Mcl-1 prevented GM-CSF-induced protection. We conclude that GM-CSF protects alveolar epithelial cells against oxidative stress-induced mitochondrial injury via the Akt pathway and its downstream components, including Mcl-1. Epithelial cell-derived GM-CSF may contribute to intrinsic defense mechanisms limiting lung injury.
Collapse
Affiliation(s)
- Anne Sturrock
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, Univ. of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | | | | | | | | | | |
Collapse
|
36
|
New insights of mitochondria reactive oxygen species generation and cell apoptosis induced by low dose photodynamic therapy. Eur J Cancer 2011; 47:2750-61. [DOI: 10.1016/j.ejca.2011.06.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 04/11/2011] [Accepted: 06/09/2011] [Indexed: 12/13/2022]
|
37
|
Oliva CR, Moellering DR, Gillespie GY, Griguer CE. Acquisition of chemoresistance in gliomas is associated with increased mitochondrial coupling and decreased ROS production. PLoS One 2011; 6:e24665. [PMID: 21931801 PMCID: PMC3170372 DOI: 10.1371/journal.pone.0024665] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 08/15/2011] [Indexed: 12/18/2022] Open
Abstract
Temozolomide (TMZ) is an alkylating agent used for treating gliomas. Chemoresistance is a severe limitation to TMZ therapy; there is a critical need to understand the underlying mechanisms that determine tumor response to TMZ. We recently reported that chemoresistance to TMZ is related to a remodeling of the entire electron transport chain, with significant increases in the activity of complexes II/III and cytochrome c oxidase (CcO). Moreover, pharmacologic and genetic manipulation of CcO reverses chemoresistance. Therefore, to test the hypothesis that TMZ-resistance arises from tighter mitochondrial coupling and decreased production of reactive oxygen species (ROS), we have assessed mitochondrial function in TMZ-sensitive and -resistant glioma cells, and in TMZ-resistant glioblastoma multiform (GBM) xenograft lines (xenolines). Maximum ADP-stimulated (state 3) rates of mitochondrial oxygen consumption were greater in TMZ-resistant cells and xenolines, and basal respiration (state 2), proton leak (state 4), and mitochondrial ROS production were significantly lower in TMZ-resistant cells. Furthermore, TMZ-resistant cells consumed less glucose and produced less lactic acid. Chemoresistant cells were insensitive to the oxidative stress induced by TMZ and hydrogen peroxide challenges, but treatment with the oxidant L-buthionine-S,R-sulfoximine increased TMZ-dependent ROS generation and reversed chemoresistance. Importantly, treatment with the antioxidant N-acetyl-cysteine inhibited TMZ-dependent ROS generation in chemosensitive cells, preventing TMZ toxicity. Finally, we found that mitochondrial DNA-depleted cells (ρ°) were resistant to TMZ and had lower intracellular ROS levels after TMZ exposure compared with parental cells. Repopulation of ρ° cells with mitochondria restored ROS production and sensitivity to TMZ. Taken together, our results indicate that chemoresistance to TMZ is linked to tighter mitochondrial coupling and low ROS production, and suggest a novel mitochondrial ROS-dependent mechanism underlying TMZ-chemoresistance in glioma. Thus, perturbation of mitochondrial functions and changes in redox status might constitute a novel strategy for sensitizing glioma cells to therapeutic approaches.
Collapse
Affiliation(s)
- Claudia R. Oliva
- Division of Neurosurgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Douglas R. Moellering
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - G. Yancey Gillespie
- Division of Neurosurgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Corinne E. Griguer
- Division of Neurosurgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
38
|
Regulation of mitochondrial metabolism: yet another facet in the biology of the oncoprotein Bcl-2. Biochem J 2011; 435:545-51. [PMID: 21486225 DOI: 10.1042/bj20101996] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The Bcl-2 (Bcl is B-cell lymphocytic-leukaemia proto-oncogene) family comprises two groups of proteins with distinct functional biology in cell-fate signalling. Bcl-2 protein was the first member to be discovered and associated with drug resistance in human lymphomas. Since then a host of other proteins such as Bcl-xL, Bcl-2A1 and Mcl-1 with similar anti-apoptotic functions have been identified. In contrast, the pro-apoptotic Bcl-2 proteins contain prototypic effector proteins such as Bax and Bak, and the BH3 (Bcl-2 homology)-only proteins comprising Bak, Bid, Bim, Puma and Noxa. A complex interplay between the association of pro-apoptotic and anti-apoptotic proteins with each other determines the sensitivity of cancer cells to drug-induced apoptosis. The canonical functional of Bcl-2 in terms of apoptosis inhibition is its ability to prevent mitochondrial permeabilization via inhibiting the translocation and oligomerization of pro-apoptotic proteins such as Bax; however, more recent evidence points to a novel mechanism of the anti-apoptotic activity of Bcl-2. Overexpression of Bcl-2 increases mitochondrial oxygen consumption and in doing so generates a slight pro-oxidant intracellular milieu, which promotes genomic instability and blocks death signalling. However, in the wake of overt oxidative stress, Bcl-2 regulates cellular redox status thereby preventing excessive build-up of ROS (reactive oxygen species), which is detrimental to cells and tissues. Taken together, the canonical and non-canonical activities of Bcl-2 imply a critical involvement of this protein in the processes of tumour initiation and progression. In the present paper we review these functionally distinct outcomes of Bcl-2 expression with implications for the chemotherapeutic management of cancers.
Collapse
|
39
|
Activation of the mitochondrial protein quality control system and actin cytoskeletal alterations in cells harbouring the MELAS mitochondrial DNA mutation. J Neurol Sci 2010; 295:46-52. [PMID: 20570288 DOI: 10.1016/j.jns.2010.05.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 05/11/2010] [Accepted: 05/17/2010] [Indexed: 01/22/2023]
Abstract
Point mutations in the mitochondrial genome are associated with a variety of metabolic disorders. The myopathy, encephalopathy, lactic acidosis, stroke-like episodes syndrome (MELAS), is most frequently associated with an A to G transition at position 3243 of the mitochondrial tRNA(Leu(UUR)) gene, and is characterized by biochemical and structural alterations of mitochondria. In the present study, we analyzed proteomic changes in an immortalized B-cell line harbouring the MELAS A3243G mutation by two-dimensional difference gel electrophoresis and immunoblot analysis. Although the cell line contained only 10% mutated mitochondrial genomes, we detected significant alterations in numerous proteins associated with the actin cytoskeleton and in nuclear-encoded subunits of mitochondrial respiratory chain complexes. Notably, mitochondrial Lon protease and Hsp60 were deregulated in MELAS cells, indicating an effect on the mitochondrial protein quality control system. By immunofluorescence microscopy, we detected mitochondrial Lon protease accumulation and changes in actin-binding proteins preferentially in MELAS cells containing numerous mitochondria with mutated genomes. Enzymatic assays revealed that Lon protease activity is increased in MELAS cell lysates. Although Lon protease has been shown to degrade misfolded proteins and to stabilize respiratory chain complexes within mitochondria, our MELAS cell line exhibited a higher sensitivity to mitochondrial stress. These findings provide novel insights into the cellular response to dysfunctional mitochondria containing mutated genomes.
Collapse
|
40
|
Hilton Z, Clements KD, Hickey AJR. Temperature sensitivity of cardiac mitochondria in intertidal and subtidal triplefin fishes. J Comp Physiol B 2010; 180:979-90. [DOI: 10.1007/s00360-010-0477-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 04/21/2010] [Accepted: 04/23/2010] [Indexed: 12/01/2022]
|
41
|
Chen ZX, Pervaiz S. Involvement of cytochrome c oxidase subunits Va and Vb in the regulation of cancer cell metabolism by Bcl-2. Cell Death Differ 2009; 17:408-20. [PMID: 19834492 DOI: 10.1038/cdd.2009.132] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Bcl-2 has been shown to promote survival of cancer cells by maintaining a slight pro-oxidant state through elevated mitochondrial respiration during basal conditions. On oxidative stress, Bcl-2 moderates mitochondrial respiration through cytochrome c oxidase (COX) activity to prevent an excessive buildup of reactive oxygen species (ROS) by-production from electron transport activities. However, the underlying molecular mechanism(s) of Bcl-2-mediated ROS regulation and its impact on carcinogenesis remain unclear. In this study, we show that Bcl-2 expression positively influences the targeting of nuclear-encoded COX Va and Vb to the mitochondria of cancer cells. In addition, evidence is presented in support of a protein-protein interaction between COX Va and Bcl-2, involving the BH2 domain of Bcl-2. Interestingly, episodes of serum withdrawal, glucose deprivation or hypoxia aimed at inducing early oxidative stress triggered Bcl-2-overexpressing cells to preserve mitochondrial levels of COX Va while depressing COX Vb, whereas the reverse was observed in mock-transfected cells. The unique manner in which Bcl-2 adjusted COX subunits during these physiological stress triggers had a profound impact on the resultant decrease in COX activity and maintenance of mitochondrial ROS levels, thus delineating a novel mechanism for the homeostatic role of Bcl-2 in the redox biology and metabolism of cancer cells.
Collapse
Affiliation(s)
- Z X Chen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | |
Collapse
|
42
|
Role of nuclear-encoded subunit Vb in the assembly and stability of cytochrome c oxidase complex: implications in mitochondrial dysfunction and ROS production. Biochem J 2009; 420:439-49. [PMID: 19338496 DOI: 10.1042/bj20090214] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
CcO (cytochrome c oxidase) is a multisubunit bigenomic protein complex which catalyses the last step of the mitochondrial electron transport chain. The nuclear-encoded subunits are thought to have roles either in regulation or in the structural stability of the enzyme. Subunit Vb is a peripheral nuclear-encoded subunit of mammalian CcO that is dramatically reduced under hypoxia. Although it has been shown to contain different ligand-binding sites and undergo modifications, its precise function is not known. In the present study we generated a cell line from RAW 264.7 murine macrophages that has a more than 80% reduced level of Vb. Functional analysis of these cells showed a loss of CcO activity, membrane potential and less ability to generate ATP. Resolution of complexes on blue native gel and two-dimensional electrophoretic analysis showed an accumulation of subcomplexes of CcO and also reduced association with supercomplexes of the electron transfer chain. Furthermore, the mitochondria from CcO Vb knock-down cells generated increased ROS (reactive oxygen species), and the cells were unable to grow on galactose-containing medium. Pulse-chase experiments suggest the role of the CcO Vb subunit in the assembly of the complex. We show for the first time the role of a peripheral, non-transmembrane subunit in the formation as well as function of the terminal CcO complex.
Collapse
|
43
|
|